Abstract
Nonalcoholic fatty liver disease (NAFLD) is the hepatic manifestation of both metabolic and inflammatory diseases and has become the leading chronic liver disease worldwide. High-fat (HF) diets promote an increased uptake and storage of free fatty acids (FFAs) and triglycerides (TGs) in hepatocytes, which initiates steatosis and induces lipotoxicity, inflammation and insulin resistance. Activation and signaling of Toll-like receptor 4 (TLR4) by FFAs induces inflammation evident in NAFLD and insulin resistance. Currently, there are no effective treatments to specifically target inflammation associated with this disease. We have established the efficacy of phenylmethimazole (C10) to prevent lipopolysaccharide and palmitate-induced TLR4 signaling. Because TLR4 is a key mediator in pro-inflammatory responses, it is a potential therapeutic target for NAFLD. Here, we show that treatment with C10 inhibits HF diet-induced inflammation in both liver and mesenteric adipose tissue measured by a decrease in mRNA levels of pro-inflammatory cytokines. Additionally, C10 treatment improves glucose tolerance and hepatic steatosis despite the development of obesity due to HF diet feeding. Administration of C10 after 16 weeks of HF diet feeding reversed glucose intolerance, hepatic inflammation, and improved hepatic steatosis. Thus, our findings establish C10 as a potential therapeutic for the treatment of NAFLD.
Introduction
Obesity is the single most important risk factor for the development of nonalcoholic fatty liver disease (NAFLD), which is the most prevalent liver disease in the western hemisphere (Lazo & Clark 2008, Bellentani et al. 2010). NAFLD, the hepatic manifestation of metabolic syndrome (Yki-Jarvinen 2014), is linked to visceral obesity (a systemic pro-inflammatory state), dyslipidemia, insulin resistance and type 2 diabetes mellitus (T2DM) (Zelber-Sagi et al. 2006). Specifically, NAFLD is a disease of excess fat accumulation in the liver of individuals with no history of alcohol abuse, which can range from benign steatosis to advanced steatohepatitis (NASH) and cirrhosis (El-Serag & Kanwal 2014). NASH is associated with increased mortality not only from vascular disease but also from complications of cirrhosis and hepatocellular cancer. Thus, targeting hepatocellular inflammation is expected to significantly prevent the progression of the disease and reduce mortality in patients with NAFLD (Younossi et al. 2011).
High-fat (HF) diets promote weight gain leading to an increase in adipose tissue mass (i.e. obesity). Simultaneously, these HF diets cause an increase in levels of circulating free fatty acids (FFAs) and triglycerides (TGs) that deposit in adipose tissue as well as in the liver and skeletal muscle (i.e. ectopic fat deposition) (Day 2002, Dowman et al. 2010). Ectopic fat deposition in the liver is the hallmark of hepatic steatosis, which is the earliest stage of NAFLD and is associated with the development of insulin resistance (Day & James 1998, Surwit et al. 1988, Xu et al. 2003, Cani et al. 2007). The ectopically deposited FFAs and TGs observed in steatosis induce a local, low-grade hepatic inflammation (Weisberg et al. 2003, Glass & Olefsky 2012) and is otherwise a benign disease at this stage (Jia et al. 2014, Sawada et al. 2014). Unfortunately, steatosis often leads to the development of NASH, which is characterized by immune cell infiltrate, hepatocyte injury and/or fibrosis.
Pathological activation and signaling of toll-like receptor 4 (TLR4) by non-immune ligands (including FFAs) and immune ligands (including gut-derived lipopolysaccharide or LPS) contribute to the inflammation present in NAFLD (Lu et al. 2008, Broering et al. 2011). TLR4 signaling is mediated via two intracellular pathways involving the myeloid differentiation primary response 88 (MyD88) or adaptor proteins translocation-associated membrane protein 1/TIR-domain-containing adapter-inducing interferon-β (TRIF) (Holland et al. 2011). In the MyD88-dependent pathway, MyD88 signals the activation of IL-1 receptor-associated kinases (IRAK4/IRAK1) and TNF receptor-associated factor (TRAF6) to activate nuclear factor κB (NFKB) and activated protein 1 (AP1) leading to the induction of pro-inflammatory cytokines (IL6, TNFA) (Paik et al. 2003, Takeda et al. 2003, Shi et al. 2006, Miura et al. 2013, O’Neill et al. 2013). In the MyD88-independent pathway, TLR4 recruits TRAF3 and receptor-interacting protein 1 (RIP1) by TRIF/toll-like receptor adaptor molecule 1 (TICAM1) to promote the downstream activation of TANK-binding kinase 1 (TBK1) and inhibitor κB kinase ε (IKKE) (Seki et al. 2001, Akira et al. 2006, Schneider et al. 2006). This molecular complex (TRIF/TICAM1/TRAF3/RIP1/TBK1/IKKE) phosphorylates interferon regulatory factor 3 (IRF3) (Fitzgerald et al. 2003). Following phosphorylation, IRF3 homodimerizes and translocates to the nucleus where it induces type 1 interferon expression (e.g. IFNB (Fitzgerald et al. 2003, Hemmi et al. 2004)). Indirectly, IRF3 interacts with NFKB and AP1 to induce IL6 and TNFA expression.
Activation of TLR4-mediated inflammation also exacerbates hepatic lipid accumulation, although the exact mechanism is still unknown. Mice deficient in TLR4 demonstrate HF diet-induced weight gain but are protected against inflammation, hepatic steatosis and insulin resistance (Shi et al. 2006, Poggi et al. 2007, Suganami et al. 2007, Tsukumo et al. 2007, Davis et al. 2008, Spruss et al. 2009, Pierre et al. 2013, Jia et al. 2014, Ferreira et al. 2015). Liver-specific TLR4-knockout (TLR4LKO) mice become obese when placed on a HF diet but remain insulin sensitive and are protected from the development of steatosis (Jia et al. 2014). The attenuation of steatosis and insulin resistance is most likely due to reduced pro-inflammatory gene expression in liver and adipose tissue of both global and liver-specific TLR4-deficient mice (Jia et al. 2014).
Even with the acknowledged epidemic of obesity and associated NAFLD, there is an overwhelming failure (1) to clinically recognize the disease in the early stages due to the lack of specific diagnostic indicators or (2) to initiate treatment as there are no effective medications which specifically attenuate the early systemic inflammatory processes of NAFLD. This leaves patients and physicians only with long-term weight loss through diet to treat NAFLD, which is effective but very difficult to sustain (Gelli et al. 2017), or bariatric surgery, which can be very effective at reducing hepatic fat content (Hannah & Harrison 2016, Schwenger et al. 2018) but can have significant associated complications (Chang et al. 2017). In light of this and studies suggesting a direct involvement of TLR4-mediated inflammation in the development of HF diet-induced hepatic steatosis and insulin resistance, there is a concerted effort directed at developing therapeutics targeting TLR4 signaling. We have developed a library of small-molecule inhibitors of inflammation that potently block TLR signaling, including FFA- and gut-derived LPS-induced TLR4 signaling (Harii et al. 2005, McCall et al. 2007, 2010, 2013, Schwartz et al. 2009, Deosarkar et al. 2014). Our lead compound, phenylmethimazole (C10), is a derivative of methimazole that inhibits inflammation resulting from TLR3 and TLR4 signaling in both immune and non-immune cells by blocking homodimerization of IRF3 and thus blocking its nuclear translocation and transcriptional activation activity (Courreges et al. 2012). Thus, we hypothesized that C10 will prevent and/or reverse HF diet-induced hepatic and adipose tissue inflammation as well as hepatic steatosis and glucose intolerance in a diet-induced obesity (DIO) mouse model.
Materials and methods
Phenylmethimazole (C10) solutions
Phenylmethimazole (C10) (Concord Biosciences, Cleveland, OH, USA) was prepared as a 200 mM stock solution in 100% (v/v) DMSO (Sigma-Aldrich) and further diluted to achieve the working concentration indicated in individual experiments.
Cell culture
Mouse hepatocyte cell line, AML-12 (ATCC), was cultured in DMEM-F12 with 0.005 mg/mL insulin, 0.005 mg/mL transferrin, 5 ng/mL selenium and 40 ng/mL dexamethasone, 10% (v/v) fetal bovine serum (Gibco) and 1% (v/v) penicillin/streptomycin. Human hepatocellular carcinoma cell line, HepG2 (ATCC) was cultured in DMEM, 10% (v/v) fetal bovine serum (Gibco) and 1% (v/v) penicillin/streptomycin (Gibco). Both cell lines were grown at 37°C with 5% CO2. A working solution of C10 (100 µM) was prepared in 0.25% DMSO (Sigma-Aldrich). For palmitate and LPS treatments, cells (cell passages <20) were incubated with 750 µM palmitate (Sigma-Aldrich) solution conjugated to FFA-free BSA (Sigma-Aldrich), 2% in serum-free culture media or complete culture media containing 10 ng/mL LPS (Sigma-Aldrich).
Mice and experimental design
This work was conducted with approval from the Ohio University Institutional Animal Care and Use Committee in accord with accepted standards of humane animal care.
Experimental procedures
Six-week-old C57BL/6J male mice were purchased from Jackson Labs and housed 4 per cage in an environment controlled for temperature (18–22°C) and humidity on a 14:10-h light/darkness cycle. Mice were allowed to acclimate for 1 week prior to diet placement and C10 and control treatments.
Prevention study
Prior to the start of the experiment, mice were randomly assigned to a treatment group: low-fat (LF) diet ((#D12450B, Research Diets Inc., New Brunswick, NJ, USA) (10% fat, 20% protein, 70% carbohydrate)) sham injection, HF diet ((#D12492, Research Diets Inc) (60% fat, 20% protein, 20% carbohydrate)) sham injection, HF diet + DMSO, HF diet + 1 mg/kg C10 in 10% DMSO and PBS. Mice received intraperitoneal (IP) injections once daily for 18 weeks. Weights were recorded weekly. Body composition (%fat, %fluid and %lean measurements) was obtained using the Bruker Minispec Whole Body Composition Analyzer (Billerica, MA, USA). An intraperitoneal glucose tolerance test (IPGTT) was performed on mice after 13 weeks on their respective diets and initiation of treatment.
Reversal study
Prior to the start of the experiment, mice were randomly assigned to a diet group: LF diet group or HF diet group. After 16 weeks on their respective diet, an IPGTT was performed to evaluate glucose tolerance in each mouse. Any mouse in the LF diet group that was glucose intolerant and any mouse in the HF diet group that were glucose tolerant were removed from the study. Inclusion/exclusion criteria were as follows: If the IPGTT curve for a HF diet-fed mouse was identical or very similar to that of the LF diet-fed group, it was excluded. Similarly, if the IPGTT curve for a LF diet-fed mouse was identical or very similar to that of the HF diet-fed group, it was excluded. One week following the IPGTTs (i.e. after 17 weeks on respective diets), mice fed the HF diet were randomly assigned to a treatment group; HF diet + sham injection, HF diet + DMSO, HF diet + 1 mg/kg C10 in 10% DMSO and PBS. Mice received once daily IP injections for 14 weeks. Weights were recorded weekly. Body composition was obtained as described earlier. Another IPGTT was performed on mice after 12 weeks of C10 or control treatments.
Intraperitoneal glucose tolerance tests (IPGTTs)
Intraperitoneal glucose tolerance tests (IPGTTs) were performed on 12-h fasted mice. Body weight and blood glucose (Freestyle Freedom Blood Glucose monitoring System, Abbott Laboratories) was measured prior to IP injection of glucose (Sigma-Aldrich) (1–2 g/kg body weight). Subsequent blood glucose measurements were performed at time 0 and at 20/30, 60, 90, 120 and 180 min post IP injection of the glucose bolus.
Histological analysis
For microscopic examination of liver morphology and steatosis, liver tissue was fixed in 10% buffered formalin for 12–24 h. Formalin-fixed tissues were dehydrated in ethanol and embedded in paraffin for hematoxylin and eosin staining. Liver sections for histological staining were cut to 5 µm. Tissue preparation for histological analysis was performed by Ohio University Heritage College of Osteopathic Medicine Histological Core Services.
Hepatic TG quantification
Hepatic TG content was evaluated using a protocol based on the Salmon and Flatt method of lipid saponification (Salmon & Flatt 1985, List et al. 2009). Glycerol concentration was plotted against absorbance. The concentration of glycerol (mg glycerol/g tissue) was calculated by multiplying the determined concentration from the equation of the graph by the dilution factor and the number 5.31. The number (5.31) was used to correct the conversion of glycerol to TG by units of glycerol (mg/dL) to units of TG (mmol/L) and to mg/g tissue. TG content in AML-12 cells was presented as a ratio of total protein determined by BCA.
Serum TG and total cholesterol
Serum TG and total cholesterol were measured using blood collected at the experimental endpoint from non-fasted mice. The commercially available colorimetric Triglyceride Quantification Assay Kit (Abcam, cat #ab65336) was performed according to the manufacturer protocol to quantify serum TG. The commercially available colorimetric Cholesterol/Cholesteryl Ester Quantitation Assay Kit (Abcam, cat #ab65359) was used according to the manufacturer protocol to measure serum total cholesterol.
Quantitative real time-PCR analysis
Total RNA was isolated from cells in culture or frozen liver and mesenteric adipose tissue using TRIzol reagent (Invitrogen, Thermo Fischer Scientific). Preparation of cDNA was achieved using the high-capacity cDNA reverse transcription kit with RNase Inhibitor (Applied Biosystems, Thermo Fischer Scientific). TaqMan (Applied Biosystems, Thermo Fischer Scientific) and SYBR Green (Bio-Rad) biochemistries was used to perform qRT-PCR to quantify gene expression according to the manufacturer’s protocol. Murine TaqMan gene expression assays used include: Ifnb (Mm00439552_s1), Adgre1 (Emr1;F4/80) (Mm00802529_m1), Il6 (Mm00446190_m1) and Gapdh (Mm99999915_g1) as the housekeeping gene. Mouse Tnfa primers were as follows: sense primer, 5′-Cgg TCC CCA AAg GGA TgA g-3′; antisense primer, 5′ CCT TgA AgA gAA CCT ggg AgT A-3′. Human TaqMan gene expression assays used include: TNFA (Hs00174128_m1), IFNB1 (Hs01077958_s1) and GAPDH (Hs02786624_g1) as the housekeeping genes.
Statistical analysis
Statistical analysis was performed using GraphPad Prism 7 for Mac. Statistical differences were determined using a one-way or two-way ANOVA followed by a Tukey–Kramer or Bonferroni test for post hoc comparison.
Results
C10 prevents FFA- and LPS-induced inflammation in both murine and human hepatocytes in culture and TG accumulation in murine hepatocytes
In previous studies, we have shown that C10 prevents palmitate- and LPS-induced pro-inflammatory cytokine expression in murine macrophages (RAW264.7 cells) and differentiated 3T3-L1 adipocytes by inhibiting TLR4 signaling, specifically by blocking transcriptional activity of IRF3 (McCall et al. 2010). In HFD-induced NAFLD, TLR4 expressed in hepatocytes is activated by both FFAs and LPS (Matsumura et al. 2000, Reyna et al. 2008). Stimulation of the MyD88-dependent pathway leads to pro-inflammatory cytokine expression, specifically Tnfa and Il6, while activation of the MyD88-independent TLR4 pathway leads to direct upregulation of type 1 interferons (Ifnb1) and indirect upregulation of Tnfa (Paik et al. 2003, Takeda et al. 2003, Shi et al. 2006, Miura et al. 2013, O’Neill et al. 2013). As indicated in Fig. 1A and B, treatment with palmitate and LPS leads to the upregulation of Ifnb1 and Tnfa in murine hepatocytes (AML-12 cells) compared to the untreated control groups and C10 prevents LPS- and palmitate-induced upregulation of Ifnb1 and Tnfa. Treatment with C10 also prevents palmitate-induced pro-inflammatory cytokine expression in HepG2 cells, a human hepatocellular carcinoma cell line (Fig. 1C). The solvent control, DMSO, is known to exhibit anti-inflammatory effects by repressing pro-inflammatory cytokine production (Elisia et al. 2016). Although DMSO had some anti-inflammatory activity, we show that C10 has a greater anti-inflammatory effect by inhibiting pro-inflammatory cytokine production when compared to the palmitate- and LPS-stimulated DMSO groups.
Inflammation is associated with enhanced hepatic de novo lipogenesis and TG accumulation (Feingold & Grunfeld 1987, Grunfeld et al. 1988, 1991, Feingold et al. 1990, 1992). Exogenous Tnfa in mice and rats has caused increased TG production and storage in the liver (Feingold & Grunfeld 1987, Feingold et al. 1990). In addition to preventing FFA- and LPS-induced pro-inflammatory cytokine expression in vitro, C10 also reduced palmitate-mediated accumulation of TG in mouse hepatocytes (AML-12 cells) (Fig. 1D).
C10 does not affect weight gain, body composition or adipose weight in HF diet-fed C57BL/6J male mice
It is already known that TLR4-deficient mice develop obesity when fed a HF diet. Thus, we were interested in the effect of C10 treatment on weight and body composition in a HF diet-induced model of obesity (DIO model). Seven-week-old C57BL6/J male mice were fed either a LF diet (10% fat) or HF diet (60% fat). Mice were dosed once daily with intraperitoneal (IP) sham injections, IP injections of DMSO (vehicle) or IP injections of 1 mg/kg C10 for 18 weeks. During the 18-week study (Prevention Study), mice were evaluated for the development of obesity by measurement of body weight and body composition. A HF diet challenge resulted in significantly more weight gain compared to the LF-fed mice (Fig. 2A). Body composition revealed increased fat mass as a percentage of total body weight in the HF-fed mice when compared to the LF-fed mice (Fig. 2B). Obesity was also assessed by adipose tissue weight. At the end of the 18-week study, HF-fed mice had increased adipose tissue weight in mesenteric, subcutaneous, epididymal and retroperitoneal depots when compared to the LF sham group (Fig. 2C). C10 and DMSO treatment had no significant effect on weight, body composition or adipose tissue weight of HF-fed mice (Fig. 2).
C10 blocks hepatic TG deposition in HF diet-fed C57BL/6J male mice
Our in vitro experiments demonstrated that C10 prevented palmitate-induced TG accumulation in mouse hepatocytes (Fig. 1D). Thus, we sought to determine if C10 could prevent hepatic TG accumulation in vivo. After 18 weeks of HF diet feeding and C10 treatment (Prevention Study), histological examination of the liver revealed the absence of steatosis in C10-treated mice when compared to livers of the HF sham and HF DMSO control mice. (Fig. 3A). To further quantify C10 inhibition of hepatic lipid accumulation, TG content was quantified biochemically which revealed increased TG content from liver samples of HF-fed mice when compared to LF-fed mice. Treatment with C10 reduced hepatic TG content compared to HF sham and HF DMSO groups (Fig. 3B). There was no difference noted among adipose depot weights of HF diet groups; however, the difference in hepatic TG content in the liver of C10-treated mice when compared to the HF controls indicate that C10 may have a localized effect on hepatic lipid metabolism. Serum TG and cholesterol levels were measured in non-fasted mice after 16 weeks of HF diet feeding (Fig. 4A and B, respectively). Total serum TG remained unchanged (Fig. 4A) while total serum cholesterol was elevated in HF diet-fed mice when compared to LF sham controls; C10 did not affect serum cholesterol levels in HF-fed mice (Fig. 4B).
C10 protects C57BL/6J male mice from HF diet-induced glucose intolerance
Ectopic fat deposition in insulin target tissues impairs the function of insulin signaling and thus impairs glucose homeostasis (i.e. induces glucose intolerance/insulin resistance). Previous in vitro studies have shown that treatment with C10 prevent palmitate-induced IRS1 serine 307 phosphorylation, a process known to mediate insulin resistance in insulin-stimulated 3T3L1 adipocytes (McCall et al. 2010). To determine the effect of C10 on glucose tolerance in HF diet-fed mice, we performed a 2-h intraperitoneal glucose tolerance test (IPGTT) 13 weeks post diet and treatment initiation. The HF-fed control animals exhibited glucose intolerance, with significantly higher glucose levels at 20, 60, 90, 120 and 180 min following glucose administration during the IPGTT compared to LF-fed mice (Fig. 5), and the area under the curve (AUC) was significantly higher in the HF-fed mice compared to LF-fed mice (Fig. 5). HF-fed mice receiving C10 treatment had improved glucose tolerance despite their obesity as compared to the HF sham and HF DMSO-treated mice (Fig. 5).
C10 prevents HF diet-induced inflammation in liver and mesenteric adipose tissue from C57BL/6J male mice
Inflammation, specifically due to cytokines and chemokines produced by FFA and gut-derived LPS activation of TLR4 signaling, leads to systemic glucose intolerance by impairing insulin signaling in target tissues including adipose and liver. Activation of the TLR4 signaling pathways leads to expression of pro-inflammatory cytokines, in particular, Tnfa and type 1 interferons (Ifnb1). Our model of HF diet feeding promotes an increase in circulating FFAs, gut-derived LPS, as well as an increase in fat deposition and accumulation in adipose tissue and the liver (Fraulob et al. 2010). Pathologic exposure of adipose and liver tissue to FFAs and gut-derived LPS activates TLR4 signaling and induces a local inflammatory tissue response marked by an increase in pro-inflammatory cytokine gene expression. We observed that palmitate treatment induces expression of Tnfa and Ifnb1 in AML-12 cells, which was inhibited by treatment with C10 (Fig. 1). Additional studies in our laboratory have shown that C10 prevents transcriptional activity of NFKB and IRF3 thereby inhibiting the upregulation of cytokine and chemokine production (McCall et al. 2010, 2013, Deosarkar et al. 2014). To determine the efficacy of C10 to prevent HF diet-induced inflammation, adipose and liver tissue were collected from the mice in this study for analysis of pro-inflammatory gene expression. In both adipose and liver tissue, there was a significant increase in Tnfa and Ifnb1 expression in our HF sham and HF DMSO groups (Fig. 6). However, as in our in vitro studies, this HF diet-mediated increase in Tnfa and Ifnb1 mRNA levels was inhibited in both liver and adipose tissues from the HF diet-fed C10-treated animals (Fig. 6A and B, respectively). Additionally, mRNA levels of F4/80, which encodes a cell surface macrophage marker remained low in adipose tissue of C10-treated mice despite HF diet feeding (Fig. 6C).
C10 reverses HF diet-induced glucose intolerance, hepatic and adipose inflammation and hepatic steatosis
The observation that C10 prevents HF diet-induced hepatic steatosis and inflammation as well as adipose inflammation and glucose intolerance in our DIO mouse model led us to question if C10 could reverse established glucose intolerance, hepatic steatosis and hepatic inflammation in these mice. To address this, male C57BL/6J mice were put on either a LF diet or a HF diet for 16 weeks after which glucose tolerance was evaluated in each mouse via IPGTT. Mice on the LF diet that were glucose tolerant remained in the study and were maintained on the LF diet for the duration of the experiment. Mice on the HF diet that were glucose intolerant remained in the study and were maintained on the HF diet for the remainder of the study. As can be seen in Figs 7 and 8, the LF-fed mice weighed significantly less than the HF-fed mice (Fig. 7A, Week 0) and the IPGTT revealed that the LF-fed mice were glucose tolerant, whereas the HF-fed mice were glucose intolerant (Fig. 8A). One week later (after 17 weeks on the diets), the glucose-intolerant HF-fed mice were randomly divided into HF sham, HF DMSO and HF C10 (1 mg/kg) treatment groups as described earlier for the ‘prevention study’ and were treated as indicated for 14 weeks. There was no difference in weights between the HF-fed treatment groups (Fig. 7A), although all HF-fed groups continued to gain weight and become more obese over the course of the 14-week treatment period (Fig. 7); however, the HF-fed C10-treated mice were now glucose tolerant (Fig. 8B), indicating that despite a continued rise in obesity, C10 reversed the glucose intolerance that was present in the mice prior to C10 treatment (Fig. 8A and B). Hepatic steatosis (Fig. 9A) was also reduced as well as total serum cholesterol (Fig. 9B). Serum TG remained unchanged (Fig. 9B). Moreover, hepatic inflammation (Tnfa, Ifnb1 and Il6) (Fig. 9C) was dramatically reduced in C10-treated mice compared to the HF controls.
Discussion
Inflammation is widely recognized as a key factor in the pathogenesis of metabolic diseases, specifically obesity-related diseases such as T2DM and NAFLD. Obesity alone is considered the most important risk factor for development of NAFLD and is the driver of inflammation in this disease that is responsible for its progression (Wild et al. 2004, Dabelea et al. 2014, Imes & Burke 2014). The major inflammatory signaling pathway in chronic inflammation in a state of obesity is TLR4 (Davis et al. 2008, Pierre et al. 2013, Jia et al. 2014, Sawada et al. 2014). TLR4 is abundantly expressed in insulin target tissues such as adipose tissue, liver and skeletal muscle and is now accepted as a key player in obesity-induced insulin resistance and T2DM (Jialal et al. 2014). Earlier studies suggested that the stimulation of TLR4 seen in obesity/insulin resistance/T2DM results from gut-derived LPS (Lassenius et al. 2011, Jayashree et al. 2014, Velloso et al. 2015); however, it is now evident that FFAs derived from HF diets can also trigger TLR4 signaling in these target tissues (Reyna et al. 2008, Kim et al. 2012) leading to NAFLD and insulin resistance.
In our model, we used a HF diet to promote the development of insulin resistance and hepatic steatosis. HF diets increase circulating levels of FFAs, which deposit in adipose tissue and other tissues such as the liver. Acceleration of hepatic FFA deposition occurs in obesity-induced NAFLD due to an increase in dietary fatty acids or lipolysis of adipose tissue. We hypothesized that C57BL/6J mice fed a HF diet would develop hepatic inflammation, steatosis and insulin resistance, which would be prevented and/or reversed with C10 treatment. Human and mouse hepatocyte cell lines demonstrated an inflammatory response when exposed to LPS and FFAs. In our in vitro system, C10 exhibited potent anti-inflammatory properties by preventing FFA- and LPS-induced pro-inflammatory cytokine expression measured by a reduction in Tnfa and Ifnb1 mRNA levels in hepatocytes in culture. Similar findings were observed in vivo as pro-inflammatory cytokine expression was also significantly reduced by C10 in liver and mesenteric adipose tissue of mice fed a HF diet. In addition to anti-inflammatory effects, C10 treatment prevented glucose intolerance (an indirect measure of insulin resistance) and hepatic steatosis in mice fed a HF diet. Inhibition of FFA-induced hepatic lipid accumulation by C10 treatment was also observed in vitro. Furthermore, and most clinically relevant, HF diet-induced insulin resistance was reversed by C10 intervention. C10-treated mice also had significantly reduced hepatic inflammation and decreased hepatic TG content, albeit the latter effect was not as dramatic as was observed in the ‘prevention study’. The modest effect of C10 on hepatic TG content in the ‘reversal study’ may be due to the fact that the HF diet used in this study induced an overwhelming amount of hepatic steatosis due to the HF diet containing 60% fat. If the C10 treatment had continued for a longer duration or the diet changed to regular chow, we anticipate this effect would be more pronounced, especially given the fact that hepatic inflammation and insulin resistance was significantly reduced. In this regard, we have previously shown that continued HF feeding after intensive insulin therapy, in this same mouse model, prevents the ‘Legacy Effect’ of early insulin treatment in new-onset T2DM (Guo et al. 2015). While not evaluated in this study, it would be of interest in future studies to see if other models of NAFLD (e.g. ob/ob or db/db mice) also respond similarly to C10 treatment.
Currently, there are no therapeutic interventions to prevent the inflammation associated with NAFLD. Because NAFLD is associated with metabolic disease and is often considered the hepatic manifestation of metabolic syndrome, pharmacological agents that target the lipid accumulation or insulin resistance component of NAFLD are used as front-line therapies. Certain anti-diabetic therapies including pioglitazone (Ratziu et al. 2008), acarbose (Chiasson et al. 2002), metformin (Haukeland et al. 2009) and possibly statins (Eslami et al. 2013) exhibit anti-inflammatory properties and are effective at treating NAFLD. However, there is a real need for novel, new classes of anti-inflammatory drugs for the prevention and treatment of the localized inflammation associated with NAFLD as the inflammation in the presence of steatosis is what leads to NASH and the more severe stages of the disease that result in death.
The pathogenesis of NAFLD is now considered to be ‘multiple-hit’ due to hepatic insults that occur in parallel, which ultimately leads to increased lipid accumulation and immune infiltration (Day & James 1998). The early stage of NAFLD (steatosis) is considered benign; however, it is now believed to be an active state of inflammation and metabolic dysfunction (Tilg & Moschen 2010, Buzzetti et al. 2016). Moreover, we now know that inflammation occurs in hepatic steatosis and thus it can be targeted by pharmacological agents before the onset of NASH. Activation of TLR4 signaling is a key mediator of HF diet-induced hepatic inflammation.
In addition to being a critical mediator of TLR4 signaling, MyD88 has recently been shown to be critical for maintaining mammalian target of rapamycin (mTOR) activation (Chang et al. 2013). Given the metabolic consequences of NAFLD (i.e. obesity, insulin resistance and T2DM), mTOR involvement is essential in light of its role in cardiovascular diseases such as atherosclerosis, coronary heart disease and stroke (Tarantino & Capone 2013, Patil & Sood 2017). Moreover, a key mechanism linking inflammation to altered glucose and lipid metabolism is that visceral adipocytes and associated macrophages produce and release copious amounts of inflammatory cytokines into both the portal and systemic vasculature, which cause insulin resistance in insulin target tissues (i.e. liver, muscle and fat). Thus, the novel findings presented herein that C10 can reverse HF diet-induced hepatic steatosis, glucose intolerance, as well as hepatic and visceral adipose inflammation, coupled with the finding that C10 inhibits Tnfa-induced Vcam1 expression and reduces monocytic cell adhesion to endothelial cells (Dagia et al. 2004), an important process in the pathogenesis of atherosclerosis and other chronic inflammatory diseases, suggests that C10 may have a more profound clinical impact than the treatment of NAFLD alone.
Declaration of interest
K D M, D J G and F S are inventors on a patent issued to Ohio University on the subject matter.
Funding
This work was supported in part by the JO Watson Endowment for Diabetes Research and 2020 Vision grant from the Ohio University Heritage College of Osteopathic Medicine, the Ohio University Heritage College of Osteopathic Medicine McCall Research Support Fund, and the John J. Kopchick Molecular and Cellular Biology (MCB)/Translational Biomedical Sciences (TBS) Fellowship Award to A P.
Author contribution statement
A P, T C, C W, J T, D G, D E B, E O L, F S, K M wrote the manuscript. A P, T C, C W, J T, D E B, E O L, F S, and K M designed and performed research and analyzed data. K M contributed new reagents/analytical tools.
Acknowledgments
The authors would like to acknowledge Debra Walter for designing the mouse TNFα primers described in 'Materials and methods' section. Additionally, we would like to acknowledge Noriko Kantake, Maria Cecilia Courreges de Benencia, Aili Guo, Megan Vogel, Alex Campolo, Kyle Mudd, Kevin Swiatek, Gregory Green, Michael Braunegg and Benjamin Niknam for their participation in the project.
References
Akira S, Uematsu S & Takeuchi O 2006 Pathogen recognition and innate immunity. Cell 124 783–801. (https://doi.org/10.1016/j.cell.2006.02.015)
Bellentani S, Scaglioni F, Marino M & Bedogni G 2010 Epidemiology of non-alcoholic fatty liver disease. Digestive Diseases 28 155–161. (https://doi.org/10.1159/000282080)
Broering R, Lu M & Schlaak JF 2011 Role of Toll-like receptors in liver health and disease. Clinical Science 121 415–426. (https://doi.org/10.1042/CS20110065)
Buzzetti E, Pinzani M & Tsochatzis EA 2016 The multiple-hit pathogenesis of non-alcoholic fatty liver disease (NAFLD). Metabolism 65 1038–1048. (https://doi.org/10.1016/j.metabol.2015.12.012)
Cani PD, Amar J, Iglesias MA, Poggi M, Knauf C, Bastelica D, Neyrinck AM, Fava F, Tuohy KM, Chabo C, et al. 2007 Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes 56 1761–1772. (https://doi.org/10.2337/db06-1491)
Chang J, Burkett PR, Borges CM, Kuchroo VK, Turka LA & Chang CH 2013 MyD88 is essential to sustain mTOR activation necessary to promote T helper 17 cell proliferation by linking IL-1 and IL-23 signaling. PNAS 110 2270–2275. (https://doi.org/10.1073/pnas.1206048110)
Chang SH, Freeman NLB, Lee JA, Stoll CRT, Calhoun AJ, Eagon JC & Colditz GA 2017 Early major complications after bariatric surgery in the USA, 2003–2014: a systematic review and meta-analysis. Obesity Reviews 19 529–537. (https://doi.org/10.1111/obr.12647)
Chiasson JL, Josse RG, Gomis R, Hanefeld M, Karasik A, Laakso M & Group S-NTR 2002 Acarbose for prevention of type 2 diabetes mellitus: the STOP-NIDDM randomised trial. Lancet 359 2072–2077. (https://doi.org/10.1016/S0140-6736(02)08905-5)
Courreges MC, Kantake N, Goetz DJ, Schwartz FL & McCall KD 2012 Phenylmethimazole blocks dsRNA-induced IRF3 nuclear translocation and homodimerization. Molecules 17 12365–12377. (https://doi.org/10.3390/molecules171012365)
Dabelea D, Mayer-Davis EJ, Saydah S, Imperatore G, Linder B, Divers J, Bell R, Badaru A, Talton JW, Crume T, et al. 2014 Prevalence of type 1 and type 2 diabetes among children and adolescents from 2001 to 2009. JAMA 311 1778–1786. (https://doi.org/10.1001/jama.2014.3201)
Dagia NM, Harii N, Meli AE, Sun X, Lewis CJ, Kohn LD & Goetz DJ 2004 Phenyl methimazole inhibits TNF-alpha-induced VCAM-1 expression in an IFN regulatory factor-1-dependent manner and reduces monocytic cell adhesion to endothelial cells. Journal of Immunology 173 2041–2049. (https://doi.org/10.4049/jimmunol.173.3.2041)
Davis JE, Gabler NK, Walker-Daniels J & Spurlock ME 2008 Tlr-4 deficiency selectively protects against obesity induced by diets high in saturated fat. Obesity 16 1248–1255. (https://doi.org/10.1038/oby.2008.210)
Day CP 2002 Pathogenesis of steatohepatitis. Best Practice and Research: Clinical Gastroenterology 16 663–678. (https://doi.org/10.1053/bega.2002.0333)
Day CP & James OF 1998 Steatohepatitis: a tale of two “hits”? Gastroenterology 114 842–845. (https://doi.org/10.1016/S0016-5085(98)70599-2)
Deosarkar SP, Bhatt P, Gillespie J, Goetz DJ & McCall KD 2014 Inhibition of LPS-induced TLR4 signaling products in murine macrophages by phenylmethimazole: an assay methodology for screening potential phenylmethimazole analogs. Drug Development Research 75 497–509. (https://doi.org/10.1002/ddr.21231)
Dowman JK, Tomlinson JW & Newsome PN 2010 Pathogenesis of non-alcoholic fatty liver disease. QJM 103 71–83. (https://doi.org/10.1093/qjmed/hcp158)
El-Serag HB & Kanwal F 2014 Epidemiology of hepatocellular carcinoma in the United States: where are we? Where do we go? Hepatology 60 1767–1775. (https://doi.org/10.1002/hep.27222)
Elisia I, Nakamura H, Lam V, Hofs E, Cederberg R, Cait J, Hughes MR, Lee L, Jia W, Adomat HH, et al. 2016 DMSO represses inflammatory cytokine production from human blood cells and reduces autoimmune arthritis. PLoS ONE 11 e0152538. (https://doi.org/10.1371/journal.pone.0152538)
Eslami L, Merat S, Malekzadeh R, Nasseri-Moghaddam S & Aramin H 2013 Statins for non-alcoholic fatty liver disease and non-alcoholic steatohepatitis. Cochrane Database of Systematic Reviews 17 CD008623. (https://doi.org/10.1002/14651858.CD008623.pub2)
Feingold KR & Grunfeld C 1987 Tumor necrosis factor-alpha stimulates hepatic lipogenesis in the rat in vivo. Journal of Clinical Investigation 80 184–190. (https://doi.org/10.1172/JCI113046)
Feingold KR, Adi S, Staprans I, Moser AH, Neese R, Verdier JA, Doerrler W & Grunfeld C 1990 Diet affects the mechanisms by which TNF stimulates hepatic triglyceride production. American Journal of Physiology 259 E177–E184.
Feingold KR, Staprans I, Memon RA, Moser AH, Shigenaga JK, Doerrler W, Dinarello CA & Grunfeld C 1992 Endotoxin rapidly induces changes in lipid metabolism that produce hypertriglyceridemia: low doses stimulate hepatic triglyceride production while high doses inhibit clearance. Journal of Lipid Research 33 1765–1776.
Ferreira DF, Fiamoncini J, Prist IH, Ariga SK, de Souza HP & de Lima TM 2015 Novel role of TLR4 in NAFLD development: modulation of metabolic enzymes expression. Biochimica et Biophysica Acta 1851 1353–1359. (https://doi.org/10.1016/j.bbalip.2015.07.002)
Fitzgerald KA, McWhirter SM, Faia KL, Rowe DC, Latz E, Golenbock DT, Coyle AJ, Liao SM & Maniatis T 2003 IKKepsilon and TBK1 are essential components of the IRF3 signaling pathway. Nature Immunology 4 491–496. (https://doi.org/10.1038/ni921)
Fraulob JC, Ogg-Diamantino R, Fernandes-Santos C, Aguila MB & Mandarim-de-Lacerda CA 2010 A mouse model of metabolic syndrome: insulin resistance, fatty liver and Non-Alcoholic Fatty Pancreas Disease (NAFPD) in C57BL/6 mice fed a high fat diet. Journal of Clinical Biochemistry and Nutrition 46 212–223. (https://doi.org/10.3164/jcbn.09-83)
Gelli C, Tarocchi M, Abenavoli L, Di Renzo L, Galli A & De Lorenzo A 2017 Effect of a counseling-supported treatment with the Mediterranean diet and physical activity on the severity of the non-alcoholic fatty liver disease. World Journal of Gastroenterology 23 3150–3162. (https://doi.org/10.3748/wjg.v23.i17.3150)
Glass CK & Olefsky JM 2012 Inflammation and lipid signaling in the etiology of insulin resistance. Cell Metabolism 15 635–645. (https://doi.org/10.1016/j.cmet.2012.04.001)
Grunfeld C, Verdier JA, Neese R, Moser AH & Feingold KR 1988 Mechanisms by which tumor necrosis factor stimulates hepatic fatty acid synthesis in vivo. Journal of Lipid Research 29 1327–1335.
Grunfeld C, Dinarello CA & Feingold KR 1991 Tumor necrosis factor-alpha, interleukin-1, and interferon alpha stimulate triglyceride synthesis in HepG2 cells. Metabolism 40 894–898. (https://doi.org/10.1016/0026-0495(91)90062-2)
Guo A, Daniels NA, Thuma J, McCall KD, Malgor R & Schwartz FL 2015 Diet is critical for prolonged glycemic control after short-term insulin treatment in high-fat diet-induced type 2 diabetic male mice. PLoS ONE 10 e0117556. (https://doi.org/10.1371/journal.pone.0117556)
Hannah WN Jr & Harrison SA 2016 Effect of weight loss, diet, exercise, and bariatric surgery on nonalcoholic fatty liver disease. Clinical Liver Disease 20 339–350. (https://doi.org/10.1016/j.cld.2015.10.008)
Harii N, Lewis CJ, Vasko V, McCall K, Benavides-Peralta U, Sun X, Ringel MD, Saji M, Giuliani C, Napolitano G, et al. 2005 Thyrocytes express a functional toll-like receptor 3: overexpression can be induced by viral infection and reversed by phenylmethimazole and is associated with Hashimoto’s autoimmune thyroiditis. Molecular Endocrinology 19 1231–1250. (https://doi.org/10.1210/me.2004-0100)
Haukeland JW, Konopski Z, Eggesbo HB, von Volkmann HL, Raschpichler G, Bjoro K, Haaland T, Loberg EM & Birkeland K 2009 Metformin in patients with non-alcoholic fatty liver disease: a randomized, controlled trial. Scandinavian Journal of Gastroenterology 44 853–860. (https://doi.org/10.1080/00365520902845268)
Hemmi H, Takeuchi O, Sato S, Yamamoto M, Kaisho T, Sanjo H, Kawai T, Hoshino K, Takeda K & Akira S 2004 The roles of two IkappaB kinase-related kinases in lipopolysaccharide and double stranded RNA signaling and viral infection. Journal of Experimental Medicine 199 1641–1650. (https://doi.org/10.1084/jem.20040520)
Holland WL, Bikman BT, Wang LP, Yuguang G, Sargent KM, Bulchand S, Knotts TA, Shui G, Clegg DJ, Wenk MR, et al. 2011 Lipid-induced insulin resistance mediated by the proinflammatory receptor TLR4 requires saturated fatty acid-induced ceramide biosynthesis in mice. Journal of Clinical Investigation 121 1858–1870. (https://doi.org/10.1172/JCI43378)
Imes CC & Burke LE 2014 The obesity epidemic: the United States as a cautionary tale for the rest of the world. Current Epidemiology Reports 1 82–88. (https://doi.org/10.1007/s40471-014-0012-6)
Jayashree B, Bibin YS, Prabhu D, Shanthirani CS, Gokulakrishnan K, Lakshmi BS, Mohan V & Balasubramanyam M 2014 Increased circulatory levels of lipopolysaccharide (LPS) and zonulin signify novel biomarkers of proinflammation in patients with type 2 diabetes. Molecular and Cellular Biochemistry 388 203–210. (https://doi.org/10.1007/s11010-013-1911-4)
Jia L, Vianna CR, Fukuda M, Berglund ED, Liu C, Tao C, Sun K, Liu T, Harper MJ, Lee CE, et al. 2014 Hepatocyte Toll-like receptor 4 regulates obesity-induced inflammation and insulin resistance. Nature Communications 5 3878. (https://doi.org/10.1038/ncomms6832)
Jialal I, Kaur H & Devaraj S 2014 Toll-like receptor status in obesity and metabolic syndrome: a translational perspective. Journal of Clinical Endocrinology and Metabolism 99 39–48. (https://doi.org/10.1210/jc.2013-3092)
Kim KA, Gu W, Lee IA, Joh EH & Kim DH 2012 High fat diet-induced gut microbiota exacerbates inflammation and obesity in mice via the TLR4 signaling pathway. PLoS ONE 7 e47713. (https://doi.org/10.1371/journal.pone.0047713)
Lassenius MI, Pietilainen KH, Kaartinen K, Pussinen PJ, Syrjanen J, Forsblom C, Porsti I, Rissanen A, Kaprio J, Mustonen J, et al. 2011 Bacterial endotoxin activity in human serum is associated with dyslipidemia, insulin resistance, obesity, and chronic inflammation. Diabetes Care 34 1809–1815. (https://doi.org/10.2337/dc10-2197)
Lazo M & Clark JM 2008 The epidemiology of nonalcoholic fatty liver disease: a global perspective. Seminars in Liver Disease 28 339–350. (https://doi.org/10.1055/s-0028-1091978)
List EO, Palmer AJ, Berryman DE, Bower B, Kelder B & Kopchick JJ 2009 Growth hormone improves body composition, fasting blood glucose, glucose tolerance and liver triacylglycerol in a mouse model of diet-induced obesity and type 2 diabetes. Diabetologia 52 1647–1655. (https://doi.org/10.1007/s00125-009-1402-z)
Lu YC, Yeh WC & Ohashi PS 2008 LPS/TLR4 signal transduction pathway. Cytokine 42 145–151. (https://doi.org/10.1016/j.cyto.2008.01.006)
Matsumura T, Ito A, Takii T, Hayashi H & Onozaki K 2000 Endotoxin and cytokine regulation of toll-like receptor (TLR) 2 and TLR4 gene expression in murine liver and hepatocytes. Journal of Interferon and Cytokine Research 20 915–921. (https://doi.org/10.1089/10799900050163299)
McCall KD, Harii N, Lewis CJ, Malgor R, Kim WB, Saji M, Kohn AD, Moon RT & Kohn LD 2007 High basal levels of functional toll-like receptor 3 (TLR3) and noncanonical Wnt5a are expressed in papillary thyroid cancer and are coordinately decreased by phenylmethimazole together with cell proliferation and migration. Endocrinology 148 4226–4237. (https://doi.org/10.1210/en.2007-0459)
McCall KD, Holliday D, Dickerson E, Wallace B, Schwartz AL, Schwartz C, Lewis CJ, Kohn LD & Schwartz FL 2010 Phenylmethimazole blocks palmitate-mediated induction of inflammatory cytokine pathways in 3T3L1 adipocytes and RAW 264.7 macrophages. Journal of Endocrinology 207 343–353. (https://doi.org/10.1677/JOE-09-0370)
McCall KD, Schmerr MJ, Thuma JR, James CB, Courreges MC, Benencia F, Malgor R & Schwartz FL 2013 Phenylmethimazole suppresses dsRNA-induced cytotoxicity and inflammatory cytokines in murine pancreatic beta cells and blocks viral acceleration of type 1 diabetes in NOD mice. Molecules 18 3841–3858. (https://doi.org/10.3390/molecules18043841)
Miura K, Yang L, van Rooijen N, Brenner DA, Ohnishi H & Seki E 2013 Toll-like receptor 2 and palmitic acid cooperatively contribute to the development of nonalcoholic steatohepatitis through inflammasome activation in mice. Hepatology 57 577–589. (https://doi.org/10.1002/hep.26081)
O’Neill LA, Golenbock D & Bowie AG 2013 The history of Toll-like receptors – redefining innate immunity. Nature Reviews Immunology 13 453–460. (https://doi.org/10.1038/nri3446)
Paik YH, Schwabe RF, Bataller R, Russo MP, Jobin C & Brenner DA 2003 Toll-like receptor 4 mediates inflammatory signaling by bacterial lipopolysaccharide in human hepatic stellate cells. Hepatology 37 1043–1055. (https://doi.org/10.1053/jhep.2003.50182)
Patil R & Sood GK 2017 Non-alcoholic fatty liver disease and cardiovascular risk. World Journal of Gastrointestinal Pathophysiology 8 51–58. (https://doi.org/10.4291/wjgp.v8.i2.51)
Pierre N, Deldicque L, Barbe C, Naslain D, Cani PD & Francaux M 2013 Toll-like receptor 4 knockout mice are protected against endoplasmic reticulum stress induced by a high-fat diet. PLoS ONE 8 e65061. (https://doi.org/10.1371/journal.pone.0065061)
Poggi M, Bastelica D, Gual P, Iglesias MA, Gremeaux T, Knauf C, Peiretti F, Verdier M, Juhan-Vague I, Tanti JF, et al. 2007 C3H/HeJ mice carrying a toll-like receptor 4 mutation are protected against the development of insulin resistance in white adipose tissue in response to a high-fat diet. Diabetologia 50 1267–1276. (https://doi.org/10.1007/s00125-007-0654-8)
Ratziu V, Giral P, Jacqueminet S, Charlotte F, Hartemann-Heurtier A, Serfaty L, Podevin P, Lacorte JM, Bernhardt C, Bruckert E, et al. 2008 Rosiglitazone for nonalcoholic steatohepatitis: one-year results of the randomized placebo-controlled Fatty Liver Improvement with Rosiglitazone Therapy (FLIRT) Trial. Gastroenterology 135 100–110. (https://doi.org/10.1053/j.gastro.2008.03.078)
Reyna SM, Ghosh S, Tantiwong P, Meka CS, Eagan P, Jenkinson CP, Cersosimo E, Defronzo RA, Coletta DK, Sriwijitkamol A, et al. 2008 Elevated toll-like receptor 4 expression and signaling in muscle from insulin-resistant subjects. Diabetes 57 2595–2602. (https://doi.org/10.2337/db08-0038)
Salmon DM & Flatt JP 1985 Effect of dietary fat content on the incidence of obesity among ad libitum fed mice. International Journal of Obesity 9 443–449.
Sawada K, Ohtake T, Hasebe T, Abe M, Tanaka H, Ikuta K, Suzuki Y, Fujiya M, Hasebe C & Kohgo Y 2014 Augmented hepatic Toll-like receptors by fatty acids trigger the pro-inflammatory state of non-alcoholic fatty liver disease in mice. Hepatology Research 44 920–934. (https://doi.org/10.1111/hepr.12199)
Schneider K, Benedict CA & Ware CF 2006 A TRAFfic cop for host defense. Nature Immunology 7 15–16. (https://doi.org/10.1038/nbib106-15)
Schwartz AL, Malgor R, Dickerson E, Weeraratna AT, Slominski A, Wortsman J, Harii N, Kohn AD, Moon RT, Schwartz FL, et al. 2009 Phenylmethimazole decreases Toll-like receptor 3 and noncanonical Wnt5a expression in pancreatic cancer and melanoma together with tumor cell growth and migration. Clinical Cancer Research 15 4114–4122. (https://doi.org/10.1158/1078-0432.CCR-09-0005)
Schwenger KJP, Fischer SE, Jackson TD, Okrainec A & Allard JP 2018 Non-alcoholic fatty liver disease in morbidly obese individuals undergoing bariatric surgery: prevalence and effect of the pre-bariatric very low calorie diet. Obesity Surgery 28 1109–1116. (https://doi.org/10.1007/s11695-017-2980-3)
Seki E, Tsutsui H, Nakano H, Tsuji N, Hoshino K, Adachi O, Adachi K, Futatsugi S, Kuida K, Takeuchi O, et al. 2001 Lipopolysaccharide-induced IL-18 secretion from murine Kupffer cells independently of myeloid differentiation factor 88 that is critically involved in induction of production of IL-12 and IL-1beta. Journal of Immunology 166 2651–2657. (https://doi.org/10.4049/jimmunol.166.4.2651)
Shi H, Kokoeva MV, Inouye K, Tzameli I, Yin H & Flier JS 2006 TLR4 links innate immunity and fatty acid-induced insulin resistance. Journal of Clinical Investigation 116 3015–3025. (https://doi.org/10.1172/JCI28898)
Spruss A, Kanuri G, Wagnerberger S, Haub S, Bischoff SC & Bergheim I 2009 Toll-like receptor 4 is involved in the development of fructose-induced hepatic steatosis in mice. Hepatology 50 1094–1104. (https://doi.org/10.1002/hep.23122)
Suganami T, Mieda T, Itoh M, Shimoda Y, Kamei Y & Ogawa Y 2007 Attenuation of obesity-induced adipose tissue inflammation in C3H/HeJ mice carrying a Toll-like receptor 4 mutation. Biochemical and Biophysical Research Communications 354 45–49. (https://doi.org/10.1016/j.bbrc.2006.12.190)
Surwit RS, Kuhn CM, Cochrane C, McCubbin JA & Feinglos MN 1988 Diet-induced type II diabetes in C57BL/6J mice. Diabetes 37 1163–1167. (https://doi.org/10.2337/diab.37.9.1163)
Takeda K, Kaisho T & Akira S 2003 Toll-like receptors. Annual Review of Immunology 21 335–376. (https://doi.org/10.1146/annurev.immunol.21.120601.141126)
Tarantino G & Capone D 2013 Inhibition of the mTOR pathway: a possible protective role in coronary artery disease. Annals of Medicine 45 348–356. (https://doi.org/10.3109/07853890.2013.770333)
Tilg H & Moschen AR 2010 Evolution of inflammation in nonalcoholic fatty liver disease: the multiple parallel hits hypothesis. Hepatology 52 1836–1846. (https://doi.org/10.1002/hep.24001)
Tsukumo DM, Carvalho-Filho MA, Carvalheira JB, Prada PO, Hirabara SM, Schenka AA, Araujo EP, Vassallo J, Curi R, Velloso LA, et al. 2007 Loss-of-function mutation in Toll-like receptor 4 prevents diet-induced obesity and insulin resistance. Diabetes 56 1986–1998. (https://doi.org/10.2337/db06-1595)
Velloso LA, Folli F & Saad MJ 2015 TLR4 at the crossroads of nutrients, gut microbiota, and metabolic inflammation. Endocrine Review 36 245–271. (https://doi.org/10.1210/er.2014-1100)
Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL & Ferrante AW Jr 2003 Obesity is associated with macrophage accumulation in adipose tissue. Journal of Clinical Investigation 112 1796–1808. (https://doi.org/10.1172/JCI200319246)
Wild S, Roglic G, Green A, Sicree R & King H 2004 Global prevalence of diabetes: estimates for the year 2000 and projections for 2030. Diabetes Care 27 1047–1053. (https://doi.org/10.2337/diacare.27.5.1047)
Xu H, Barnes GT, Yang Q, Tan G, Yang D, Chou CJ, Sole J, Nichols A, Ross JS, Tartaglia LA, et al. 2003 Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. Journal of Clinical Investigation 112 1821–1830. (https://doi.org/10.1172/JCI200319451)
Yki-Jarvinen H 2014 Non-alcoholic fatty liver disease as a cause and a consequence of metabolic syndrome. Lancet Diabetes and Endocrinology 2 901–910. (https://doi.org/10.1016/S2213-8587(14)70032-4)
Younossi ZM, Stepanova M, Rafiq N, Makhlouf H, Younoszai Z, Agrawal R & Goodman Z 2011 Pathologic criteria for nonalcoholic steatohepatitis: interprotocol agreement and ability to predict liver-related mortality. Hepatology 53 1874–1882. (https://doi.org/10.1002/hep.24268)
Zelber-Sagi S, Nitzan-Kaluski D, Halpern Z & Oren R 2006 Prevalence of primary non-alcoholic fatty liver disease in a population-based study and its association with biochemical and anthropometric measures. Liver International 26 856–863. (https://doi.org/10.1111/j.1478-3231.2006.01311.x)